Skip to main content
Integrative Medicine: A Clinician's Journal logoLink to Integrative Medicine: A Clinician's Journal
. 2014 Aug;13(4):35–43.

Mitochondrial Dysfunction and Chronic Disease: Treatment With Natural Supplements

Garth L Nicolson 1,
PMCID: PMC4566449  PMID: 26770107

Abstract

Loss of function in mitochondria, the key organelle responsible for cellular energy production, can result in the excess fatigue and other symptoms that are common complaints in almost every chronic disease. At the molecular level, a reduction in mitochondrial function occurs as a result of the following changes: (1) a loss of maintenance of the electrical and chemical transmembrane potential of the inner mitochondrial membrane, (2) alterations in the function of the electron transport chain, or (3) a reduction in the transport of critical metabolites into mitochondria. In turn, these changes result in a reduced efficiency of oxidative phosphorylation and a reduction in production of adenosine-5′-triphosphate (ATP). Several components of this system require routine replacement, and this need can be facilitated with natural supplements. Clinical trials have shown the utility of using oral replacement supplements, such as l-carnitine, alpha-lipoic acid (α-lipoic acid [1,2-dithiolane-3-pentanoic acid]), coenzyme Q10 (CoQ10 [ubiquinone]), reduced nicotinamide adenine dinucleotide (NADH), membrane phospholipids, and other supplements. Combinations of these supplements can reduce significantly the fatigue and other symptoms associated with chronic disease and can naturally restore mitochondrial function, even in long-term patients with intractable fatigue.


Mitochondrial dysfunction, characterized by a loss of efficiency in the electron transport chain and reductions in the synthesis of high-energy molecules, such as adenosine-5′-triphosphate (ATP), is a characteristic of aging, and essentially, of all chronic diseases.14 These diseases include neurodegenerative diseases, such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, and Friedreich’s ataxia1,2,4,5; cardiovascular diseases, such as atherosclerosis and other heart and vascular conditions6,7; diabetes and metabolic syndrome810; autoimmune diseases, such as multiple sclerosis, systemic lupus erythematosus, and type 1 diabetes1114; neurobehavioral and psychiatric diseases, such as autism spectrum disorders, schizophrenia, and bipolar and mood disorders1519; gastrointestinal disorders20,21; fatiguing illnesses, such as chronic fatigue syndrome and Gulf War illnesses2224; musculoskeletal diseases, such as fibromyalgia and skeletal muscle hypertrophy/atrophy2527; cancer28,29; and chronic infections.30,31

It is well known among researchers that mitochondrial genetic or primary mitochondrial disorders contribute to mitochondrial dysfunction and secondary or acquired degenerative disorders.32 This review will concentrate on nongenetic or acquired mechanisms that could explain mitochondrial dysfunction and their replacement treatment with natural supplements and combinations of natural supplements, including vitamins, minerals, enzyme cofactors, antioxidants, metabolites, transporters, membrane-type phospholipids, and other natural supplements.

Mitochondrial Molecular Dysfunction

Mitochondrial dysfunction arises from an inadequate number of mitochondria, an inability to provide necessary substrates to mitochondria, or a dysfunction in their electron transport and ATP-synthesis machinery. The number and functional status of mitochondria in a cell can be changed by (1) fusion of partially dysfunctional mitochondria and mixing of their undamaged components to improve overall function, (2) the generation of entirely new mitochondria (fission), and (3) the removal and complete degradation of dysfunctional mitochondria (mitophagy).33 These events are controlled by complex cellular processes that sense the deterioration of mitochondria, such as the depolarization of mitochondrial membranes or the activation of certain transcription pathways.34,35

The ability of cells to produce almost all high-energy molecules such as ATP is directly related to the ability of mitochondria to (1) convert the energy of metabolites to reduced nicotinamide adenine dinucleotide (NADH) and (2) transfer electrons from NADH to the electron transport chain and eventually to molecular oxygen while pumping protons from the mitochondrial matrix across the inner mitochondrial membrane to the intermembrane space. This process creates a transmembrane proton gradient (Δp) and an electrochemical gradient (Δψm) across the mitochondrial inner membrane.36,37 The transmembrane potential created by the proton gradient then uses ATP synthase to flow protons back across the inner mitochondrial membrane and employs the energy from this process to drive adenosine diphosphate (ADP) phosphorylation to ATP.36,38

A consequence of the electron transport process is the production of reactive oxygen species (ROS), highly reactive free radicals that are produced as a by-product of oxidative phosphorylation. The main sources of ROS and the related reactive nitrogen species (RNS) are mitochondria, and these free radicals can damage cellular lipids, proteins, and DNA.3941 However, some mechanisms can neutralize ROS/RNS; dismutase enzymes and antioxidants can control excess amounts of ROS/RNS.42,43 In addition to creation of ROS/RNS, the electron transport process can induce uncoupling proteins, resulting in a controlled leak of protons back across the proton gradient of the inner mitochondrial membrane into the mitochondrial matrix.36,37 This leak results in reduced ATP production while it still consumes excess oxygen.43

In the presence of a controlled proton leak, excess oxygen consumption and the resulting ROS production can result in inappropriate damage to mitochondrial membrane lipids,41,42 such as the very ROS/RNS-sensitive cardiolipin, an inner mitochondrial membrane phospholipid.44 Oxidative damage to the cardiolipin and other membrane phospholipids in the inner mitochondrial membrane can result in increased proton and ion leakage back across the inner membrane into the mitochondrial matrix and partial loss of the electrochemical gradient. Cardiolipin is also an important component of the electron transport chain, providing stability for the cytochrome/enzyme complexes in the inner mitochondrial membrane.44 Once adversely damaged by ROS/RNS, oxidized cardiolipin instigates loss of electron-transport function.45

Cellular antioxidant defenses usually maintain ROS/RNS levels at concentrations that prevent excess oxidation of cellular molecules.4648 Cellular antioxidant defenses that are endogenous are mediated by glutathione peroxidase, catalase, and superoxide dismutase, among other enzymes.48,49 Also, some dietary antioxidants with a low molecular weight can affect antioxidant status.50,51 Some of these dietary antioxidants have been used as natural preventive agents to shift the excess concentrations of oxidative molecules down to physiological levels that can be maintained by endogenous antioxidant systems.52

Mitochondrial Dysfunction and Fatigue

Mitochondrial dysfunction is directly related to excess fatigue. Fatigue is considered a multidimensional sensation that is perceived to be a loss of overall energy and an inability to perform even simple tasks without exertion.53,54 Although mild fatigue can be caused by a number of conditions, including depression and other psychological conditions, moderate to severe fatigue involves cellular energy systems.53,54 At the cellular level, moderate to severe fatigue is related to loss of mitochondrial function and diminished production of ATP.5456 Intractable fatigue lasting more than 6 months that is not reversed by sleep (chronic fatigue) is the most common complaint of patients seeking general medical care.53,57 Chronic fatigue is also an important secondary condition in many clinical diagnoses, often preceding patients’ primary diagnoses.57,58

As a result of aging and chronic diseases, oxidative damage to mitochondrial membranes impairs mitochondrial function.5961 As an example, individuals with chronic fatigue syndrome present with evidence of oxidative damage to DNA and lipids,61,62 such as oxidized blood markers63 and oxidized membrane lipids,64 that is indicative of excess oxidative stress. These individuals also have sustained, elevated levels of peroxynitrite caused by excess nitric oxide, which can also result in lipid peroxidation and loss of mitochondrial function as well as changes in cytokine levels that exert a positive feedback on nitric oxide production.65

Natural Supplements and Mitochondrial Dysfunction

A number of natural supplements have been used to treat nonpsychological fatigue and mitochondrial dysfunction.32,54,66 These supplements include those containing vitamins, minerals, antioxidants, metabolites, enzyme inhibitors and cofactors, mitochondrial transporters, herbs, and membrane phospholipids (Table 1).32 Although several natural supplements have been used to reduce fatigue, few are considered truly effective.67 This article will discuss some of the most promising supplements and conclude with combinations of specific supplements that have been used to treat intractable chronic fatigue and improve mitochondrial function.

Table 1.

An Incomplete List of Ingredients/Agents That Medical Practitioners Have Used or Suggested for Treatment of Mitochondrial Dysfunctiona

Category Examples
Vitamins Vitamins C, D and E, thiamine, riboflavin
Minerals Magnesium, calcium, phosphate
Lipids Membrane phospholipids, unsaturated fatty acids
Metabolites Creatine, pyruvate
Cofactors CoQ10, α-lipoic acid, NADH, nicotinic acid
Transporters l-Carnitine, membrane phospholipids
Antioxidants CoQ10, α-lipoic acid, NADH, glutathione
Enzyme inhibitors α-Lipoic acid, dichloroacetate
Herbs Curcumin, schisandrin

Abbreviations: NADH = reduced nicotinamide adenine dinucleotide; CoQ10 = coenzyme Q10 (ubiquinone); α-lipoic acid = alpha-lipoic acid.

a

Modified from Kerr.32

α-Lipoic Acid

Alpha-lipoic acid (α-lipoic acid [1,2-dithiolane-3-pentanoic acid]) is a potent antioxidant, transition metalion chelator, redox transcription regulator, and anti-inflammatory agent.68 It acts as a critical cofactor in mitochondrial α-keto acid dehydrogenases, and thus it is important in mitochondrial, oxidative-decarboxylation reactions.69,70 Clinically, α-lipoic acid has been used as an oral supplement in the treatment of complications associated with diabetes mellitus, and according to a review by Shay et al,70 it has been shown to bring about improvements in various diabetes-associated neuropathies, inflammation, and vascular health. In model cellular systems, these effects have been attributed mainly to α-lipoic acid having signal-transduction effects on gene regulation and on glucose uptake and metabolism as well as its antioxidant effects.71

As a result of aging and in many chronic diseases, certain sphingolipids—especially ceramides, and in particular, short-chain ceramides—accumulate in mitochondria due to hydrolysis of sphingomyelin by sphingomyelinase, and eventually, this accumulation retards electron transport activity.72,73 Ceramide accumulation in mitochondria is particularly damaging to cardiac tissue. In aging rodents, α-lipoic acid has been used to lower ceramide levels in vascular endothelial cells of cardiac muscle through inhibition of sphingomyelinase activity, resulting in restoration of mitochondrial glutathione levels and increasing electron transport function.74

As mentioned above, in diabetes, α-lipoic acid has been used extensively to reduce diabetic complications, such as sensorimotor polyneuropathies.75 One 4-year, blinded study demonstrated that some neuropathic impairments improved significantly on α-lipoic acid (but not nerve conduction attributes), showing the antioxidant’s clinical utility and the safety of long-term treatment with α-lipoic acid for diabetic patients.76

Given as an oral supplement, α-lipoic acid is rarely present in tissues above micromolar levels; thus, it is unlikely to be directly involved as an important primary cellular antioxidant.70 However, its ability to increase cellular glutathione levels is an important antioxidant property, and this increase is accomplished by regulating glutathione synthesis and thus ameliorating oxidative stress.77 This antioxidant can affect the regulation of the nuclear transcription factor NF-κΒ, and, thus, it can cause widespread transcriptional effects, resulting in the attenuation of production of free radicals and cytotoxic cytokines.78

As a transition metal chelater, α-lipoic acid can remove excess copper, iron, and other metals that are involved in chronic diseases, such as hemochromatosis, end-stage renal failure, and Alzheimer’s and Parkinson’s diseases, and it is a potential therapeutic agent for prevention or mitigation of heavy metal poisoning.68 It also improves cognitive function and mitochondrial function, suggesting a link between oxidative damage to mitochondria and cognition.79 The use of α-lipoic acid for chronic fatigue has not yet been studied in controlled clinical trials, but its widespread use as a safe supplement (usually 200–600 mg/d)70 to support mitochondrial function and reduce oxidative stress has justified its incorporation into various supplement mixtures.70,76,78

l-Carnitine

l-carnitine (3-hydroxy-4-Ν-trimethylaminobutyrate) is a naturally occurring fatty acid transporter found in all species of mammals. It is directly involved in the transport of fatty acids into the mitochondrial matrix for subsequent β-oxidation, but it also functions in removal of excess acyl groups from the body and in the modulation of intracellular coenzyme A (CoA) homeostatasis.80,81 Because of its importance in fatty acid oxidation and CoA and acyl-CoA homeostatasis, l-carnitine is usually maintained within relatively narrow concentration limits; thus, dietary supplementation is important to maintain optional l-carnitine concentrations within cells.81 Indeed, l-carnitine deficiency disorders are associated with reduced mitochondrial function, insulin resistance, and coronary artery disease.8284

The important role of l-carnitine in mitochondrial metabolism has spurred the use of l-carnitine supplements to potentially improve physical performance.85 The rationale is that increased reliance on fat as the principle substrate for energy production during extreme exercise should reduce the need for carbohydrates and delay the depletion of carbohydrate stores and that these changes should increase overall energy production and reduce exercise-induced fatigue. Transport of fatty acids into mitochondria also requires increased levels of l-carnitine, and, thus, indicates a need for dietary supplementation of l-carnitine. However, studies have shown that increasing oral l-carnitine supplementation, even for 2 to 3 weeks prior to extreme exercise, did not increase carnitine content in skeletal muscle. Therefore, it is unlikely that this supplementation alters muscle metabolism during extreme exercise.86,87

l-carnitine supplementation has been successfully used in clinical disorders that are characterized by low concentrations of l-carnitine or impaired fatty acid oxidation, such as diabetes, sepsis, renal disease, and cardiomyopathy.80 For example, in a small study of 18 individuals with congestive heart failure and 12 placebo controls, propionyl-l-carnitine supplementation resulted in increased peak heart rate (mean of 12%), exercise capacity (mean of 21%), and peak oxygen consumption (mean of 45%) in the treatment group.88

An important antiaging use of l-carnitine has been to increase the rate of mitochondrial oxidative phosphorylation that naturally declines as a result of aging. This decline impairs energy production while it increases production of ROS/RNS. Feeding old rats acetyl-l-carnitine was found to reverse age-related decreases in l-carnitine levels while it increased fatty acid metabolism. It also reversed the age-related decline in cellular glutathione levels and improved the complex IV activity of muscle mitochondria.89

Although dietary supplementation with l-carnitine and its various derivatives appears to be safe (doses up to 2 g/d)90 and potentially useful in increasing mitochondrial function, researchers have not performed the necessary multiple clinical trials to show its effectiveness in most age-related chronic illnesses (other than diabetes and cardiovascular diseases). One exception was a randomized, controlled clinical trial on 70 centenarians who were treated with l-carnitine for 6 months. These participants were generally found to have muscle weakness, decreasing mental health, impaired mobility, and poor endurance. By the end of the study, the treated group showed significant improvements in physical fatigue, mental fatigue, and fatigue severity. They also showed reductions in total fat mass, increased muscle mass, and an increased capacity for physical and cognitive activity through reduced fatigue and improved cognitive function.91 Other trials on alcoholism, hepatic encephalopathy, coronary heart diseases, Peyronie’s disease, cerebral ischemia, and infertility indicated that administration of l-carnitine can have positive effects on signs and symptoms of these conditions.90

Coenzyme Q10

Coenzyme Q10 (CoQ10 [ubiquinone]) is a key cofactor and component of the mitochondrial electron transport chain and one of the most widely used natural supplements.32,92 It is also a strong antioxidant in its reduced form, and it can affect the expression of certain genes involved in cell signaling, metabolism, and transport.92,93 However, the main role of CoQ10 is its involvement in the transfer of electrons along the multiple complexes of the mitochondrial electron transport chain.92,94 Clinically, it has been used in doses up to 1200 mg per day, but most studies used lower doses.92

Because CoQ10 is an important component of the mitochondrial oxidative phosphorylation system, its supplementation in individuals with reduced levels should result in increased energy production and reduced fatigue.92,94 A systematic review of the effects of CoQ10 in physical exercise, hypertension, and heart failure by Rosenfeldt et al95 revealed that 6 out of 11 published studies showed modest improvements in exercise capacity in the participants given dietary CoQ10. In 8 of the studies, which examined the effects of CoQ10 on hypertension, a mean decrease occurred in blood pressure: systolic, a mean decline of 16 mm Hg; and diastolic, a mean decline of 10 mm Hg. In the review, 9 randomized trials that examined the use of CoQ10 in participants with heart failure showed nonsignificant trends toward increased injection fraction and reduced mortality. Rosenfeldt et al95 performed their own 3-month, randomized, placebo-controlled trial on the effects of oral CoQ10 in 35 patients with heart failure and found that participants in the CoQ10 arm, but not in the control arm, showed significant improvements in symptoms.95 The study also showed a trend toward improvements in mean exercise times.95

The effects of administration of oral CoQ10 during physical exercise have also been examined. In a blinded, crossover trial, 17 healthy participants received CoQ10 or a placebo for 8 days, and their performance was then evaluated twice at fixed workloads on a bicycle ergometer for 2 hours, with a 4-hour rest between the tests.96 The participants on CoQ10 were able to achieve higher work outputs and had less fatigue sensations, and their need for a recovery period was alleviated compared to the placebo group.96

Clinically, CoQ10 has been used to reduce symptoms and progression in various neurodegenerative diseases.92,94 In studies using Alzheimer’s disease models, CoQ10 administration significantly delayed brain atrophy and typical β-amyloid-plaque pathology.97,98 In a randomized, placebo-controlled, 16-week clinical trial on 98 Alzheimer’s participants who took an oral mixture of CoQ10, vitamins C and E, and α-lipoic acid, the test arm showed significant reductions in oxidative-stress markers but did not show significant changes in cerebrospinal fluid (CSF) markers related to β-amyloid or tau pathology.99

In Parkinson’s disease, individuals generally show increased oxidized-to-total CoQ10 ratios and significant increases in markers of oxidative damage in the CSF, which can be partially reversed with CoQ10 administration.100 In individuals with early Huntington’s disease, the Huntington Study Group’s trial showed that CoQ10 administration for 30 months slowed the usual decline in total functional capacity, but the differences did not reach statistical significance.101 Finally, in a multicenter, placebo-controlled, phase II trial with amyotrophic lateral-sclerosis patients, CoQ10 did not significantly modify functional decline over a 9-month period,102 and Mathews et al103 did not find CoQ10 plus several vitamins to be effective in individuals with genetic mitochondrial diseases.

Nicotinamide Adenine Dinucleotide

NADH functions as a cellular redox cofactor in over 200 cellular redox reactions and as substrate for certain enzymes.104,105 Humans universally require NADH, and its deficiency results in pellagra, which is characterized by dermatitis, diarrhea, dementia, and eventually death.104 In the mitochondria, NADH delivers electrons from metabolite hydrolysis to the electron transport chain, but in its reduced form, it can also act as a strong antioxidant. The usual route of dietary supplementation has historically been via NADH precursors, such as niacin, nicotinic acid, or nicotinamide, but recently, microcarriers have been used to stabilize oral NADH so that it can be directly ingested in small doses and absorbed in the gastrointestinal system. This supplementation turns out to be more effective than large oral doses of NADH, as in some studies that used up to 50 mg/kg/d. At that size of dose, the NADH is prone to oxidation and degradation, and such supplementation is generally considered to be ineffective.106

In neurodegenerative diseases, oxidative damage is extensive,1,2 and various mitochondrial antioxidants have been used to treat disease and delay progression.16,32 In Alzheimer’s disease, one study showed that stabilized oral NADH could improve cognitive functioning and dementia106; however, another clinical trial showed no evidence of improvements in cognition or dementia using oral NADH.107 In a controlled trial with 26 Alzheimer’s participants who were given stabilized NADH or placebo for 6 months, Dermin et al108 found that that the test group had significantly better performance scores than the placebo group in verbal fluency and visual construction and showed a trend toward increased performance on abstract verbal reasoning. However, the study provided no evidence of better performance for measures of attention or memory or on scores of dementia severity.

Stabilized, oral NADH has also been used to ameliorate the symptoms of Parkinson’s disease. In a preliminary, open-label clinical trial, Birkmayer et al109 examined the effects of IV and oral NADH in over 800 individuals with Parkinson’s disease. They found that 19.3% of participants showed a 30% to 50% decrease in disability; 58.8% had moderate (10%–30%) improvement; and 21.8% did not respond to the therapy (P < .01). Younger patients with a shorter duration of disease had a much better chance of responding and showing more significant improvements than older patients or patients with a longer duration of disease. The oral form was comparable to IV NADH in its effects.109 When they repeated this type of trial, however, Dizdar et al110 did not find statistically significant improvements in Parkinson’s disease rating scores in participants treated with NADH, and differences were also not found in CSF clinical markers associated with Parkinson’s disease severity.

Stabilized, oral NADH has also been used to reduce symptoms in patients with chronic fatigue. One such study on individuals with chronic fatigue syndrome was designed to treat participants with stabilized, oral NADH or placebo for 4 weeks in a crossover trial.111 Of these participants, 8 of 26 (30.7%) responded positively to the microencapsulated NADH compared with 2 of 26 (8%) in the placebo arm (P < .05).111 Clearly an effect occurred but only in a subset of participants in the trial; thus, these results were not considered significant by others.112 A clinical trial that compared oral, stabilized NADH to psychological/nutritional therapy for 31 individuals with chronic fatigue syndrome revealed that stabilized NADH alone reduced fatigue in the first 4 months of a 12-month trial. After the first 4 months, however, symptom scores were similar in the 2 arms of the trial.113 In another study, stabilized NADH was given orally for 2 months to treat individuals with chronic fatigue syndrome.114 This treatment resulted in decreases in anxiety and in maximum heart rate after a stress test, but Alegre et al114 found few or no differences in the functional impact on fatigue, quality of life, sleep quality, exercise capacity, or functional reserve. Thus stabilized NADH alone has shown mixed results in various diseases and disorders, and not every patient responded to the oral, stabilized supplement.

Membrane Phospholipids

The dietary replacement of mitochondrial membrane phospholipids (lipid replacement therapy [LRT]) using food-derived molecules to remove damaged, mainly oxidized, membrane lipids in mitochondria and other cellular organelles has proved very effective at increasing mitochondrial function and reducing fatigue.10,23,54,55 To some degree, antioxidant supplements can reduce ROS/RNS levels and prevent some oxidation of mitochondrial membrane phospholipids, but antioxidants alone cannot repair the damage already done to cells, and in particular, to cells’ mitochondrial inner membranes.29,55,115

The use of oral membrane phospholipids plus antioxidants in doses ranging from 500 to 2000 mg per day has been effective in the treatment of certain clinical conditions, such as chronic fatigue and fatiguing illnesses.55,67,115117 LRT results in the actual replacement of damaged membrane phospholipids with undamaged (unoxidized) lipids to ensure proper function of cellular and particularly mitochondrial membranes.

Oral membrane phospholipids can increase mitochondrial function and decrease fatigue in chronic fatigue syndrome, fibromyalgia syndrome, and other fatiguing conditions, including natural aging (Table 2). For example, a mixture of membrane phospholipids and vitamins (Propax with NT Factor) was used by Ellithorpe et al117 in a study on aging individuals with severe chronic fatigue and was found to reduce their fatigue by 40.5% in 8 weeks. In these studies, fatigue was monitored using the Piper Fatigue Scale (PFS) to measure clinical fatigue and quality of life.58 In a subsequent crossover study, the effects of membrane phospholipids on fatigue and mitochondrial function in patients with moderate-to-severe, chronic fatigue was initiated.55 Oral administration of NT Factor for 12 weeks resulted in a 35.5% reduction in fatigue and 26.8% increase in mitochondrial function, whereas after a 12-week washout period, fatigue increased and mitochondrial function decreased back toward control levels.55 Similar findings on fatigue reduction were observed in individuals with chronic fatigue syndrome and fibromyalgia syndrome who were given oral membrane phospholipids (NT Factor).116 Using a new formulation of NT Factor plus vitamins, minerals, and other supplements in individuals with moderate chronic fatigue resulted in a 36.8% reduction in fatigue within 1 week (Table 2).118

Table 2.

Oral Membrane Phospholipid Supplementation and Fatigue in Chronically Ill Patientsa

Participants/Patients n Avg Age Avg Time on LRT PFS Fatigue Reduction (%) Reference
Chronic fatigue 34 50.3 8 wk 40.5c Ellithorpe et al117
Aging, chronic fatigue 20 68.9 12 wk 35.5b Agadjanyan et al55
Chronic fatigue syndrome (and/or fibromyalgia syndrome) 15 44.8 8 wk 43.1b Nicolson & Ellithorpe116
Aging, fatigue 67 57.3 1 wk 36.8b Nicolson et al118
Chronic illnesses 58 55.0 8 wk 30.7b Nicolson et al119

Abbreviations: Avg = average; LRT = lipid replacement therapy; PFS = Piper Fatigue Scale.

a

Modified from Nicolson and Settineri.54

b

P < .001 compared to no supplement.

c

P < .0001 compared to no supplement.

Combination Oral Supplement to Reduce Fatigue

In a 2-month trial of the treatment of long-term intractable fatigue in patients with a variety of diagnoses, the author and several colleagues combined membrane phospholipids (2000 mg/d), CoQ10 (35 mg/d), microencapsulated NADH (35 mg/d), l-carnitine (160 mg/d), α-ketoglutaric acid (180 mg/d), and other nutrients into an oral supplement (ATP Fuel) to treat fatigue and mitochondrial dysfunction.119,120 The 58 participants in this trial had moderate-to-severe, intractable fatigue for an average of >17 years and had been to an average of >15 practitioners without resolution of their fatigue. The study included 30 individuals with chronic fatigue syndrome; 17 with chronic Lyme disease; 16 with other fatiguing illnesses, including fibromyalgia syndrome and Gulf War illness; 4 with autoimmune disease, including rheumatoid arthritis; 2 with cancer; and 2 with diabetes. These patients had tried many drugs and supplements (average >35) to reduce their fatigue without success.

Participants in this trial took the combination LRT supplement (ATP Fuel) for 8 weeks, and fatigue was scored using the PFS.119 The PFS is a validated instrument that measures four dimensions of subjective fatigue: behavioral/severity, affective/meaning, sensory, and cognitive/mood.58 The study used the instrument to calculate the four subscale or dimensional scores and the total fatigue scores. Participants had initial, total, mean PFS fatigue scores of 7.51 ± 0.29, and after 8 weeks of supplements, the mean scores improved to 5.21 ± 0.28, or a 30.7% reduction in fatigue (t test, P < .0001; Wilcoxon signed-rank, P < .0001).119

PFS scores can be further dissected into 4 subcategories: (1) the behavior/severity subcategory, which deals with completing tasks, socializing, and engaging in sexual and other activities and with intensity or degree of fatigue; (2) the affective/meaning subcategory, which determines whether an individual finds the fatigue/tiredness to be pleasant or unpleasant, agreeable or disagreeable, protective or destructive, and normal or abnormal; (3) the sensory subcategory, which determines whether an individual feels strong or weak, awake or sleepy, refreshed or tired, and energetic or unenergetic; and (4) the cognitive/mood subcategory, which assesses whether an individual feels relaxed or tense, exhilarated or depressed, and able or unable to concentrate, remember, and think clearly. In the study being discussed in this section, all of these subcategories showed significant reductions by the end of the 8-week trial (P < .0001), indicating that significant improvements occurred for all subcategories of fatigue. For example, a 30.7% reduction (P < .0001) in severity/behavior of fatigue occurred, indicating a significant reduction in the intensity of fatigue and a significant increase in the ability to complete tasks, socialize, and engage in sexual and other activities. Also, a 28.0% improvement (P < .0001) occurred in mood and cognitive ability, such as the ability to concentrate, remember, and think clearly.119

Regression Analysis of Fatigue Data

To determine the trends in fatigue reduction as the result of participants’ use of the combination LRT supplement (membrane phospholipids, CoQ10, NADH, l-carnitine, α-ketoglutaric acid, and other ingredients) over the time of the trial, the author and colleagues conducted regression analyses of the data to determine if results were (1) consistent, (2) occurred with a high degree of confidence, and (3) could predict further reductions in fatigue.119 The regression analysis of overall fatigue and of each of the subcategories of fatigue indicated significant and consistent downward trends in the fatigue data, suggesting that further reductions in fatigue would have been likely if the trial had been continued. The regression R2 values for the various subgroups were (1) behavior/severity, 0.956; (2) affective meaning, 0.960; (3) sensory, 0.950; and (4) cognitive/mood, 0.980. Regression analysis of the overall fatigue yielded R2 = 0.960. This finding indicated that a high level of confidence and reproducibility existed in the downward trends in all fatigue data. The combination LRT supplement was safe, and no safety issues came up during the trial.119 Examination of scores from individuals with chronic fatigue syndrome, Lyme disease, or other diagnosis categories did not reveal major differences in overall fatigue or its reduction by the combination supplement.119,120

Summary

Oral natural supplements containing membrane phospholipids, CoQ10, microencapsulated NADH, l-carnitine, α-lipoic acid, and other nutrients can help restore mitochondrial function and reduce intractable fatigue in patients with chronic illnesses. The combination of these supplements can result in a safe and effective method to reduce fatigue and help restore quality of life.

Footnotes

Author Disclosure Statement

The author has received no financial benefit from and has no conflict of interest regarding the products discussed in this article.

References

  • 1.Swerdlow RH. Brain aging, Alzheimer’s disease, and mitochondria. Biochim Biophys Acta. 2011;1812(12):1630–1639. doi: 10.1016/j.bbadis.2011.08.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Reddy PH. Mitochondrial medicine for aging and neurodegenerative diseases. Neuromolecular Med. 2008;10(4):291–315. doi: 10.1007/s12017-008-8044-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Green DR, Galluzzi L, Kroemer G. Mitochondria and the autophagy-inflammation-cell death axis in organismal aging. Science. 2011;333(6046):1109–1112. doi: 10.1126/science.1201940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Reddy PH, Reddy TP. Mitochondria as a therapeutic target for aging and neurodegenerative diseases. Curr Alzheimer Res. 2011;8(4):393–409. doi: 10.2174/156720511795745401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Karbowski M, Neutzner A. Neurodegeneration as a consequence of failed mitochondrial maintenance. Acta Neuropathol. 2012;123(2):157–171. doi: 10.1007/s00401-011-0921-0. [DOI] [PubMed] [Google Scholar]
  • 6.Victor VM, Apostolova N, Herance R, Hernandez-Mijares A, Rocha M. Oxidative stress and mitochondrial dysfunction in atherosclerosis: mitochondria-targeted antioxidants as potential therapy. Curr Med Chem. 2009;16(35):4654–4667. doi: 10.2174/092986709789878265. [DOI] [PubMed] [Google Scholar]
  • 7.Limongelli G, Masarone D, D’Alessandro R, Elliott PM. Mitochondrial diseases and the heart: an overview of molecular basis, diagnosis, treatment and clinical course. Future Cardiol. 2012;8(1):71–88. doi: 10.2217/fca.11.79. [DOI] [PubMed] [Google Scholar]
  • 8.Ma ZA, Zhao Z, Turk J. Mitochondrial dysfunction and beta-cell failure in type 2 diabetes mellitus. Exp Diabetes Res. 2012;2012:703538. doi: 10.1155/2012/703538. doi:10.1155/2012/703538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Joseph AM, Joanisse DR, Baillot RG, Hood DA. Mitochondrial dysregulation in the pathogenesis of diabetes: potential for mitochondrial biogenesis-mediated interventions. Exp Diabetes Res. 2012;2012:642038. doi: 10.1155/2012/642038. doi:10.1155/2012/642038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Nicolson GL. Metabolic syndrome and mitochondrial function: molecular replacement and antioxidant supplements to prevent membrane peroxidation and restore mitochondrial function. J Cell Biochem. 2007;100(6):1352–1369. doi: 10.1002/jcb.21247. [DOI] [PubMed] [Google Scholar]
  • 11.Ghafourifar P, Mousavizadeh K, Parihar MS, Nazarewicz RR, Parihar A, Zenebe WJ. Mitochondria in multiple sclerosis. Front Biosci. 2008 Jan;13:3116–3126. doi: 10.2741/2913. [DOI] [PubMed] [Google Scholar]
  • 12.Mao P, Reddy PH. Is multiple sclerosis a mitochondrial disease? Biochim Biophys Acta. 2010;1802(1):66–79. doi: 10.1016/j.bbadis.2009.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Fernandez D, Perl A. Metabolic control of T cell activation and death in SLE. Autoimmun Rev. 2009;8(3):184–189. doi: 10.1016/j.autrev.2008.07.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Maiese K, Morhan SD, Chong ZZ. Oxidative stress biology and cell injury during type 1 and type 2 diabetes mellitus. Curr Neurovasc Res. 2007;4(1):63–71. doi: 10.2174/156720207779940653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Rossignol DA, Frye RE. Mitochondrial dysfunction in autism spectrum disorders: a systematic review and meta-analysis. Mol Psychiatry. 2012;17(3):290–314. doi: 10.1038/mp.2010.136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Palmieri L, Persico AM. Mitochondrial dysfunction in autism spectrum disorders: cause or effect? Biochim Biophys Acta. 2010;1797(6–7):1130–1137. doi: 10.1016/j.bbabio.2010.04.018. [DOI] [PubMed] [Google Scholar]
  • 17.Prince JA, Harro J, Blennow K, Gottfries CG, Oreland L. Putamen mitochondrial energy metabolism is highly correlated to emotional and intellectual impairment in schizophrenics. Neuropsychopharmacology. 2000;22(3):284–292. doi: 10.1016/S0893-133X(99)00111-6. [DOI] [PubMed] [Google Scholar]
  • 18.Marazziti D, Baroni S, Piccheti M, et al. Psychiatric disorders and mitochondrial dysfunctions. Eur Rev Med Pharmacol Sci. 2012;16(2):270–275. [PubMed] [Google Scholar]
  • 19.Konradi C, Eaton M, MacDonald ML, Walsh J, Benes FM, Heckers S. Molecular evidence for mitochondrial dysfunction in bipolar disorder. Arch Gen Psychiatry. 2004;61(3):300–308. doi: 10.1001/archpsyc.61.3.300. [DOI] [PubMed] [Google Scholar]
  • 20.Chitkara DK, Nurko S, Shoffner JM, Buie T, Flores A. Abnormalities in gastrointestinal motility are associated with diseases of oxidative phosphorylation in children. Am J Gastroenterol. 2003;98(4):871–877. doi: 10.1111/j.1572-0241.2003.07385.x. [DOI] [PubMed] [Google Scholar]
  • 21.Di Donato S. Multisystem manifestations of mitochondrial disorders. J Neurol. 2009;256(5):693–710. doi: 10.1007/s00415-009-5028-3. [DOI] [PubMed] [Google Scholar]
  • 22.Norheim KB, Jonsson G, Omdal R. Biological mechanisms of chronic fatigue. Rheumatology (Oxford) 2011;50(6):1009–1018. doi: 10.1093/rheumatology/keq454. [DOI] [PubMed] [Google Scholar]
  • 23.Nicolson GL, Nicolson NL, Berns P, Nasralla MY, Haier J, Nass M. Gulf War Illnesses: chemical, radiological and biological exposures resulting in chronic fatiguing illnesses can be identified and treated. J Chronic Fatigue Syndr. 2003;11(1):135–154. [Google Scholar]
  • 24.Myhill S, Booth NE, McLaren-Howard J. Chronic fatigue syndrome and mitochondrial dysfunction. Int J Clin Exp Med. 2009;2(1):1–16. [PMC free article] [PubMed] [Google Scholar]
  • 25.Cordero MD, de Miguel M, Carmona-Lopez I, Bonal P, Campa F, Moreno-Fernandez AM. Oxidative stress and mitochondrial dysfunction in fibromyalgia. Neuro Endocrinol Lett. 2010;31(2):169–173. [PubMed] [Google Scholar]
  • 26.Rabinovich RA, Vilaro J. Structural and functional changes of peripheral muscles in chronic obstructive pulmonary disease patients. Curr Opin Pulm Med. 2010;16(2):123–133. doi: 10.1097/MCP.0b013e328336438d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Breeding PC, Russell NC, Nicolson GL. Integrative model of chronically activated immune-hormonal pathways important in the generation of fibromyalgia. Br J Med Pract. 2012;5(3):a524–a534. [Google Scholar]
  • 28.Sotgia F, Martinez-Outschoorn UE, Lisanti MP. Mitochondrial oxidative stress drives tumor progression and metastasis: should we use antioxidants as a key component of cancer treatment and prevention? BMC Med. 2011;9:62–67. doi: 10.1186/1741-7015-9-62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Nicolson GL. Lipid replacement therapy: a nutraceutical approach for reducing cancer-associated fatigue and the adverse effects of cancer therapy while restoring mitochondrial function. Cancer Metastasis Rev. 2010;29(3):543–552. doi: 10.1007/s10555-010-9245-0. [DOI] [PubMed] [Google Scholar]
  • 30.Gabridge MG. Metabolic consequences of Mycoplasma pneumoniae infection. Isr J Med Sci. 1987;23(6):574–579. [PubMed] [Google Scholar]
  • 31.Ashida H, Mimuro H, Ogawa M, et al. Cell death and infection: a double-edged sword for host and pathogen survival. J Cell Biol. 2011;195(6):931–942. doi: 10.1083/jcb.201108081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Kerr DS. Treatment of mitochondrial electron transport chain disorders: a review of clinical trials over the past decade. Mol Genet Metab. 2010;99(3):246–255. doi: 10.1016/j.ymgme.2009.11.005. [DOI] [PubMed] [Google Scholar]
  • 33.Twig G, Shirihai OS. The interplay between mitochondrial dynamics and mitophagy. Antioxid Redox Signal. 2011;14(10):1939–1951. doi: 10.1089/ars.2010.3779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Priault M, Salin B, Schaeffer J, Vallette FM, di Rago JP, Martinou JC. Impairing the bioenergetic status and the biogenesis of mitochondria triggers mitophagy in yeast. Cell Death Differ. 2005;12(12):1613–1621. doi: 10.1038/sj.cdd.4401697. [DOI] [PubMed] [Google Scholar]
  • 35.Lee J, Giordano S, Zhang J. Autophagy, mitochondria and oxidative stress: cross-talk and redox signaling. Biochem J. 2012;441(2):523–540. doi: 10.1042/BJ20111451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Rich PR, Marechal A. The mitochondrial respiratory chain. Essays Biochem. 2010;47:1–23. doi: 10.1042/bse0470001. [DOI] [PubMed] [Google Scholar]
  • 37.Nicholls DG. Mitochondrial ion circuits. Essays Biochem. 2010;47:25–35. doi: 10.1042/bse0470025. [DOI] [PubMed] [Google Scholar]
  • 38.Divakaruni AS, Brand MD. The regulation and physiology of mitochondrial proton leak. Physiology (Bethesda) 2011;26(3):192–205. doi: 10.1152/physiol.00046.2010. [DOI] [PubMed] [Google Scholar]
  • 39.Richter C, Park JW, Ames BN. Normal oxidative damage to mitochondrial and nuclear DNA is extensive. Proc Nat Acad Sci USA. 1998;85(17):6465–6467. doi: 10.1073/pnas.85.17.6465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Stadtman E. Introduction to serial reviews on oxidatively modified proteins in aging and disease. Free Radic Biol Med. 2002;32(9):789. doi: 10.1016/s0891-5849(02)00780-3. [DOI] [PubMed] [Google Scholar]
  • 41.Spector AA, Yorek MA. Membrane lipid composition and cellular function. J Lipid Res. 1985;26(9):1015–1035. [PubMed] [Google Scholar]
  • 42.Spiteller G. Is lipid peroxidation of polyunsaturated acids the only source of free radicals that induce aging and age-related diseases? Rejuvenation Res. 2010;13(1):91–103. doi: 10.1089/rej.2009.0934. [DOI] [PubMed] [Google Scholar]
  • 43.Duchen MR, Szabadkai G. Roles of mitochondria in human disease. Essays Biochem. 2010;47:115–137. doi: 10.1042/bse0470115. [DOI] [PubMed] [Google Scholar]
  • 44.Chicco AJ, Sparagna GC. Role of cardiolipin alterations in mitochondrial dysfunction and disease. Am J Physiol Cell Physiol. 2007;292(1):C33–C44. doi: 10.1152/ajpcell.00243.2006. [DOI] [PubMed] [Google Scholar]
  • 45.Houtkooper RH, Vaz FM. Cardiolipin, the heart of mitochondrial metabolism. Cell Mol Life Sci. 2008;65(16):2493–2506. doi: 10.1007/s00018-008-8030-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Barber DA, Harris SR. Oxygen free radicals and antioxidants: a review. Am Pharm. 1994;NS34(9):26–35. doi: 10.1016/s0160-3450(15)30310-x. [DOI] [PubMed] [Google Scholar]
  • 47.Sun Y. Free radicals, antioxidant enzymes, and carcinogenesis. Free Radic Biol Med. 1990;8(6):583–599. doi: 10.1016/0891-5849(90)90156-d. [DOI] [PubMed] [Google Scholar]
  • 48.Fridovich I. Superoxide radical and superoxide dismutases. Annu Rev Biochem. 1995;64:97–112. doi: 10.1146/annurev.bi.64.070195.000525. [DOI] [PubMed] [Google Scholar]
  • 49.Chandra Jagetia G, Rajanikant GK, Rao SK, Shrinath Baliga M. Alteration in the glutathione, glutathione peroxidase, superoxide dismutase and lipid peroxidation by ascorbic acid in the skin of mice exposed to fractionated gamma radiation. Clin Chim Acta. 2003;332(1–2):111–121. doi: 10.1016/s0009-8981(03)00132-3. [DOI] [PubMed] [Google Scholar]
  • 50.Aeschbach R, Loliger J, Scott BC, et al. Antioxidant actions of thymol, carvacrol, 6-gingerol, zingerone and hydroxytyrosol. Food Chem Toxicol. 1994;32(1):31–36. doi: 10.1016/0278-6915(84)90033-4. [DOI] [PubMed] [Google Scholar]
  • 51.Schwartz JL. The dual roles of nutrients as antioxidants and prooxidants: their effects on tumor cell growth. J Nutr. 1996;126(4) suppl:1221S–1227S. doi: 10.1093/jn/126.suppl_4.1221S. [DOI] [PubMed] [Google Scholar]
  • 52.Prasad KN, Cole WC, Kumar B, Prasad KC. Scientific rationale for using high-dose multiple micronutrients as an adjunct to standard and experimental cancer therapies. J Am Coll Nutr. 2001;20(5) suppl:450S–453S. doi: 10.1080/07315724.2001.10719184. [DOI] [PubMed] [Google Scholar]
  • 53.Kroenke K, Wood DR, Mangelsdorff AD, Meier NJ, Powell JB. Chronic fatigue in primary care: prevalence, patient characteristics, and outcome. JAMA. 1988;260(7):929–934. [PubMed] [Google Scholar]
  • 54.Nicolson GL, Settineri R. Lipid Replacement Therapy: a functional food approach with new formulations for reducing cellular oxidative damage, cancer-associated fatigue and the adverse effects of cancer therapy. Funct Foods Health Dis. 2011;1(4):135–160. [Google Scholar]
  • 55.Agadjanyan M, Vasilevko V, Ghochikyan A, et al. Nutritional supplement (NTFactor) restores mitochondrial function and reduces moderately severe fatigue in aged subjects. J Chronic Fatigue Syndr. 2003;11(3):23–26. [Google Scholar]
  • 56.Booth NE, Myhill S, McLaren-Howard J. Mitochondrial dysfunction and the pathophysiology of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) Int J Clin Esp Med. 2012;5(3):208–220. [PMC free article] [PubMed] [Google Scholar]
  • 57.Morrison JD. Fatigue as a presenting complaint in family practice. J Family Pract. 1980;10(5):795–801. [PubMed] [Google Scholar]
  • 58.Piper BF, Linsey AM, Dodd MJ. Fatigue mechanisms in cancer patients: developing nursing theory. Oncol Nurs Forum. 1987;14(6):17–23. [PubMed] [Google Scholar]
  • 59.Wei YH, Lee HC. Oxidative stress, mitochondrial DNA mutation, and impairment of antioxidant enzymes in aging. Exp Biol Med (Maywood) 2002;227(9):671–682. doi: 10.1177/153537020222700901. [DOI] [PubMed] [Google Scholar]
  • 60.Huang H, Manton KG. The role of oxidative damage in mitochondria during aging: a review. Front Biosci. 2004 May;9:1100–1117. doi: 10.2741/1298. [DOI] [PubMed] [Google Scholar]
  • 61.Logan AC, Wong C. Chronic fatigue syndrome: oxidative stress and dietary modifications. Altern Med Rev. 2001;6(5):450–459. [PubMed] [Google Scholar]
  • 62.Manuel y Keenoy B, Moorkens G, Vertommen J, De Leeuw I. Antioxidant status and lipoprotein peroxidation in chronic fatigue syndrome. Life Sci. 2001;68(17):2037–2049. doi: 10.1016/s0024-3205(01)01001-3. [DOI] [PubMed] [Google Scholar]
  • 63.Richards RS, Roberts TK, McGregor NR, Dunstan RH, Butt HL. Blood parameters indicative of oxidative stress are associated with symptom expression in chronic fatigue syndrome. Redox Rep. 2000;5(1):35–41. doi: 10.1179/rer.2000.5.1.35. [DOI] [PubMed] [Google Scholar]
  • 64.Fulle S, Mecocci P, Fano G, et al. Specific oxidative alterations in vastus lateralis muscle of patients with the diagnosis of chronic fatigue syndrome. Free Radic Biol Med. 2000;29(12):1252–1259. doi: 10.1016/s0891-5849(00)00419-6. [DOI] [PubMed] [Google Scholar]
  • 65.Pall ML. Elevated, sustained peroxynitrite levels as the cause of chronic fatigue syndrome. Med Hypotheses. 2000;54(1):115–125. doi: 10.1054/mehy.1998.0825. [DOI] [PubMed] [Google Scholar]
  • 66.Dimauro S, Rustin P. A critical approach to the therapy of mitochondrial respiratory chain and oxidative phosphorylation diseases. Biochim Biophys Acta. 2009;1792(12):1159–1167. doi: 10.1016/j.bbadis.2008.10.015. [DOI] [PubMed] [Google Scholar]
  • 67.Chambers D, Bagnall AM, Hempel S, Forbes C. Interventions for the treatment management and rehabilitation of patients with chronic fatigue syndrome/myalgic encepthalomyelitis: an updated systematic review. J R Soc Med. 2006;99(10):506–520. doi: 10.1258/jrsm.99.10.506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Smith AR, Shenvi SV, Widlansky M, Suh JH, Hagen TM. Lipoic acid as a potential therapy for chronic diseases associated with oxidative stress. Curr Med Chem. 2004;11(9):1135–1146. doi: 10.2174/0929867043365387. [DOI] [PubMed] [Google Scholar]
  • 69.Maczurek A, Hager K, Kenklies M, et al. Lipoic acid as an anti-inflammatory and neuroprotective treatment for Alzheimer’s disease. Adv Drug Deliv Rev. 2008;60(13–14):1463–1470. doi: 10.1016/j.addr.2008.04.015. [DOI] [PubMed] [Google Scholar]
  • 70.Shay KP, Moreau RF, Smith EJ, Smith AR, Hagen TM. Alpha-lipoic acid as a dietary supplement: molecular mechanisms and therapeutic potential. Biochim Biophys Acta. 2009;1790(10):1149–1160. doi: 10.1016/j.bbagen.2009.07.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Henriksen EJ, Jacob S, Streeper RS, Fogt DL, Hokama JY, Tritschler HJ. Stimulation by alpha-lipoic acid of glucose transport activity in skeletal muscle of lean and obese Zucker rats. Life Sci. 1997;61(8):805–812. doi: 10.1016/s0024-3205(97)00562-6. [DOI] [PubMed] [Google Scholar]
  • 72.Gudz TI, Tserng KY, Hoppel CL. Direct inhibition of mitochondrial respiratory chain complex III by cell-permeable ceramide. J Biol Chem. 1997;272(39):24154–24158. doi: 10.1074/jbc.272.39.24154. [DOI] [PubMed] [Google Scholar]
  • 73.Di Paola M, Cocco T, Lorusso M. Ceramide interaction with the respiratory chain of heart mitochondria. Biochemistry. 2000;39(22):6660–6668. doi: 10.1021/bi9924415. [DOI] [PubMed] [Google Scholar]
  • 74.Monette JS, Gomez LA, Moreau RF, et al. (R)-α-Lipoic acid treatment restores ceramide balance in aging rat cardiac mitochondria. Pharmacol Res. 2011;63(1):23–29. doi: 10.1016/j.phrs.2010.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Ziegler D, Nowak H, Kempler P, Vargha P, Low PA. Treatment of symptomatic diabetic polyneuropathy with the antioxidant alpha-lipoic acid: a meta-analysis. Diabet Med. 2004;21(2):114–121. doi: 10.1111/j.1464-5491.2004.01109.x. [DOI] [PubMed] [Google Scholar]
  • 76.Ziegler D, Low PA, Litchy WJ, et al. Efficacy and safety of antioxidant treatment with α-lipoic acid over 4 years in diabetic polyneuropathy: the NATHAN 1 trial. Diabetes Care. 2011;34(9):2054–2060. doi: 10.2337/dc11-0503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Yoshida K, Hirokawa J, Tagami S, Kawakami Y, Urata Y, Kondo T. Weakened cellular scavenging activity against oxidative stress in diabetes mellitus: regulation of glutathione synthesis and efflux. Diabetologia. 1995;38(2):201–210. doi: 10.1007/BF00400095. [DOI] [PubMed] [Google Scholar]
  • 78.Goraca A, Huk-Kolega H, Piechota A, Kleniewska P, Ciejka E, Skibska B. Lipoic acid—biological activity and therapeutic potential. Pharmacol Rep. 2011;63(4):849–858. doi: 10.1016/s1734-1140(11)70600-4. [DOI] [PubMed] [Google Scholar]
  • 79.Head E, Nukala VN, Fenoglio KA, Muggenburg BA, Cotman CW, Sullivan PG. Effects of age, dietary and behavioral enrichment on brain mitochondria in a canine model of human aging. Exp Neurol. 2009;220(1):171–176. doi: 10.1016/j.expneurol.2009.08.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Marcovina SM, Sirtori C, Peracino A, et al. Translating the basic knowledge of mitochondrial functions to metabolic therapy: role of L-carnitine. Transl Res. 2013;161(2):73–84. doi: 10.1016/j.trsl.2012.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Reuter SE, Evans AM. Carnitine and acylcarnitines: pharmacokinetic, pharmacological and clinical aspects. Clin Pharmacokinet. 2012;51(9):553–572. doi: 10.1007/BF03261931. [DOI] [PubMed] [Google Scholar]
  • 82.Newgard CB, An J, Bain JR, et al. A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metab. 2009;9(4):311–326. doi: 10.1016/j.cmet.2009.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Koves TR, Ussher JR, Noland RC, et al. Mitochondrial overload and incomplete fatty acid oxidation contribute to skeletal muscle insulin resistance. Cell Metab. 2008;7(1):45–56. doi: 10.1016/j.cmet.2007.10.013. [DOI] [PubMed] [Google Scholar]
  • 84.Shah SH, Hauser ER, Bain JR, et al. High heritability of metabolomic profiles in families burdened with premature cardiovascular disease. Mol Syst Biol. 2009;5:258. doi: 10.1038/msb.2009.11. doi:10.1038/msb.2009.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Spriet LL, Perry CG, Talanian JL. Legal pre-event nutritional supplements to assist energy metabolism. Essays Biochem. 2008;44:27–43. doi: 10.1042/BSE0440027. [DOI] [PubMed] [Google Scholar]
  • 86.Brass EP. Supplemental carnitine and exercise. Am J Clin Nutr. 2000;72(2) suppl:618S–623S. doi: 10.1093/ajcn/72.2.618S. [DOI] [PubMed] [Google Scholar]
  • 87.Wachter S, Vogt M, Kreis R, et al. Long-term administration of L-carnitine to humans: effect on skeletal muscle carnitine content and physical performance. Clin Chim Acta. 2002;318(1–2):51–61. doi: 10.1016/s0009-8981(01)00804-x. [DOI] [PubMed] [Google Scholar]
  • 88.Anand I, Chandrashekhan Y, De Giuli F, et al. Acute and chronic effects of propionyl-L-carnitine on the hemodynamics, exercise capacity and hormones in patients with congestive heart failure. Cardiovasc Drugs Ther. 1998;12(3):291–299. doi: 10.1023/a:1007721917561. [DOI] [PubMed] [Google Scholar]
  • 89.Hagen TM, Moreau R, Suh JH, Visioli F. Mitochondrial decay in the aging rat heart: evidence for improvement by dietary supplementation with acetyl-L-carnitine and/or lipoic acid. Ann NY Acad Sci. 2002 Apr;959:491–507. doi: 10.1111/j.1749-6632.2002.tb02119.x. [DOI] [PubMed] [Google Scholar]
  • 90.Acetyl-L-carnitine: monograph. Altern Med Rev. 2010;15(1):76–83. [PubMed] [Google Scholar]
  • 91.Malaguarnera M, Cammalleri L, Gargante MP, Vacante M, Colonna V, Motta M. L-carnitine treatment reduces severity of physical and mental fatigue and increases cognitive functions in centenarians: a randomized and controlled clinical trial. Am J Clin Nutr. 2007;86(6):1738–1744. doi: 10.1093/ajcn/86.5.1738. [DOI] [PubMed] [Google Scholar]
  • 92.Littarru GP, Tiano L. Clinical aspects of coenzyme Q10: an update. Nutrition. 2010;26(3):250–254. doi: 10.1016/j.nut.2009.08.008. [DOI] [PubMed] [Google Scholar]
  • 93.Groneberg DA, Kindermann B, Althammer M, et al. Coenzyme Q10 affects expression of genes involved in cell signalling, metabolism and transport in CaCo-2 cells. Int J Biochem Cell Biol. 2005;37(6):1208–1218. doi: 10.1016/j.biocel.2004.11.017. [DOI] [PubMed] [Google Scholar]
  • 94.Orsucci D, Mancuso M, Ienco EC, LoGerfo A, Siciliano G. Targeting mitochondrial dysfunction and neurodegeneration by means of coenzyme Q10 and its analogues. Curr Med Chem. 2011;18(26):4053–4064. doi: 10.2174/092986711796957257. [DOI] [PubMed] [Google Scholar]
  • 95.Rosenfeldt F, Hilton D, Pepe S, Krum H. Systematic review of effect of coenzyme Q10 in physical exercise, hypertension and heart failure. Biofactors. 2003;18(1–4):91–100. doi: 10.1002/biof.5520180211. [DOI] [PubMed] [Google Scholar]
  • 96.Mizuno K, Tanaka M, Nozaki S, et al. Antifatigue effects of coenzyme Q10 during physical fatigue. Nutrition. 2008;24(4):293–299. doi: 10.1016/j.nut.2007.12.007. [DOI] [PubMed] [Google Scholar]
  • 97.Li G, Jack CR, Yang XF, Yang ES. Diet supplement CoQ10 delays brain atrophy in aged transgenic mice with mutations in the amyloid precursor protein: an in vivo volume MRI study. Biofactors. 2008;32(1–4):169–178. doi: 10.1002/biof.5520320120. [DOI] [PubMed] [Google Scholar]
  • 98.Yang X, Dai G, Li G, Yang ES. Coenzyme Q10 reduces beta-amyloid plaque in an APP/PS1 transgenic mouse model of Alzheimer’s disease. J Mol Neurosci. 2010;41(1):110–113. doi: 10.1007/s12031-009-9297-1. [DOI] [PubMed] [Google Scholar]
  • 99.Galasko DR, Peskind E, Clark CM, et al. Alzheimer’s Disease Cooperative Study. Antioxidants for Alzheimer’s disease: a randomized clinical trial with cerebrospinal fluid biomarker measures. Arch Neurol. 2012;69(7):836–841. doi: 10.1001/archneurol.2012.85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Isobe C, Abe T, Terayama Y. Levels of reduced and oxidized coenzyme Q10 and 8-hydroxy-2’-deoxyguanosine in the cerebrospinal fluid of patients with living Parkinson’s disease demonstrates that mitochondrial oxidative damage and/or oxidative DNA damage contributes to the neurodegenerative process. Neurosci Lett. 2010;469(1):159–163. doi: 10.1016/j.neulet.2009.11.065. [DOI] [PubMed] [Google Scholar]
  • 101.Huntington Study Group. A randomized, placebo-controlled trial of coenzyme Q10 and remacemide in Huntington’s disease. Neurology. 2001;57(3):397–404. doi: 10.1212/wnl.57.3.397. [DOI] [PubMed] [Google Scholar]
  • 102.Kaufmann P, Thompson JL, Levy G, et al. QALS Study Group. Phase II trial of CoQ10 for ALS finds insufficient evidence to justify phase III. Ann Neurol. 2009;66(2):235–244. doi: 10.1002/ana.21743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Matthews PM, Ford B, Dandurand RJ, et al. Coenzyme Q10 with multiple vitamins is generally ineffective in treatment of mitochondrial disease. Neurology. 1993;43(5):884–890. doi: 10.1212/wnl.43.5.884. [DOI] [PubMed] [Google Scholar]
  • 104.Penberthy WT. Nicotinamide adenine dinucleotide biology and disease. Curr Pharm Des. 2009;15(1):1–2. doi: 10.2174/138161209787185779. [DOI] [PubMed] [Google Scholar]
  • 105.Kirkland JB. Niacin status, NAD distribution and ADP-ribose metabolism. Curr Pharm Des. 2009;15(1):3–11. doi: 10.2174/138161209787185823. [DOI] [PubMed] [Google Scholar]
  • 106.Birkmayer JG. Coenzyme nicotinamide adenine dinucleotide: new therapeutic approach for improving dementia of the Alzheimer type. Ann Clin Lab Sci. 1996;26(1):1–9. [PubMed] [Google Scholar]
  • 107.Rainer M, Kraxberger E, Haushofer M, Mucke HA, Jellinger KA. No evidence for cognitive improvement from oral nicotinamide adenine dinucleotide (NADH) in dementia. J Neural Transm. 2000;107(12):1475–1481. doi: 10.1007/s007020070011. [DOI] [PubMed] [Google Scholar]
  • 108.Demarin V, Podobnik SS, Storga-Tomic D, Kay G. Treatment of Alzheimer’s disease with stabilized oral nicotinamide adenine dinucleotide: a randomized, double-blind study. Drugs Exp Clin Res. 2004;30(1):27–33. [PubMed] [Google Scholar]
  • 109.Birkmayer JG, Vrecko C, Volc D, Birkmayer W. Nicotinamide adenine dinucleotide (NADH)—a new therapeutic approach to Parkinson’s disease: comparison of oral and parenteral application. Acta Neurol Scand Suppl. 1993;146:32–35. [PubMed] [Google Scholar]
  • 110.Dizdar N, Kagedal B, Lindvall B. Treatment of Parkinson’s disease with NADH. Acta Neurol Scand. 1994;90(5):345–347. doi: 10.1111/j.1600-0404.1994.tb02735.x. [DOI] [PubMed] [Google Scholar]
  • 111.Forsyth LM, Preuss HG, MacDowell AL, Chiazze L, Jr, Birkmayer GD, Bellanti JA. Therapeutic effects of oral NADH on the symptoms of patients with chronic fatigue syndrome. Ann Allergy Asthma Immunol. 1999;82(2):185–191. doi: 10.1016/S1081-1206(10)62595-1. [DOI] [PubMed] [Google Scholar]
  • 112.Colquhoun D, Senn S. Is NADH effective in the treatment of chronic fatigue syndrome? Ann Allergy Asthma Immunol. 2000;84(6):639–640. doi: 10.1016/s1081-1206(10)62421-0. [DOI] [PubMed] [Google Scholar]
  • 113.Santaella ML, Font I, Disdier OM. Comparison of oral nicotinamide adenine dinucleotide (NADH) versus conventional therapy for chronic fatigue syndrome. P R Health Sci J. 2004;23(2):89–93. [PubMed] [Google Scholar]
  • 114.Alegre J, Rosés JM, Javierre C, Ruiz-Baqués A, Segundo MJ, de Sevilla TF. Nicotinamide adenine dinucleotide (NADH) in patients with chronic fatigue syndrome [in Spanish] Rev Clin Esp. 2010;210(6):284–288. doi: 10.1016/j.rce.2009.09.015. [DOI] [PubMed] [Google Scholar]
  • 115.Nicolson GL. Lipid replacement as an adjunct to therapy for chronic fatigue, anti-aging and restoration of mitochondrial function. J Am Nutraceutical Assoc. 2003;6(3):22–28. [Google Scholar]
  • 116.Nicolson GL, Ellithorpe R. Lipid replacement and antioxidant nutritional therapy for restoring mitochondrial function and reducing fatigue in chronic fatigue syndrome and other fatiguing illnesses. J Chronic Fatigue Syndr. 2006;13(1):57–68. [Google Scholar]
  • 117.Ellithorpe RR, Settineri RA, Nicolson GL. Pilot study: reduction of fatigue by use of a dietary supplement containing glycophospholipids. J Am Nutraceutical Assoc. 2003;6(1):23–28. [Google Scholar]
  • 118.Nicolson GL, Ellithorpe RR, Ayson-Mitchell C, Jacques B, Settineri R. Lipid replacement therapy with a glycophospholipid-antioxidant-vitamin formulation significantly reduces fatigue within one week. J Am Nutraceutical Assoc. 2010;13(1):10–14. [Google Scholar]
  • 119.Nicolson GL, Settineri R, Ellithorpe R. Lipid replacement therapy with a glycophospholipid formulation with NADH and CoQ10 significantly reduces fatigue in intractable chronic fatiguing illnesses and chronic Lyme disease patients. Int J Clin Med. 2012;3(3):163–170. [Google Scholar]
  • 120.Nicolson GL, Settineri R, Ellithorpe R. Glycophospholipid formulation with NADH and CoQ10 significantly reduces intractable fatigue in Western blot-positive chronic Lyme disease patients: preliminary report. Funct Foods Health Dis. 2012;2(3):35–47. [Google Scholar]

Articles from Integrative Medicine: A Clinician's Journal are provided here courtesy of InnoVision Media

RESOURCES